Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 113
Filtrar
1.
Sci Immunol ; 4(31)2019 01 04.
Artículo en Inglés | MEDLINE | ID: mdl-30610013

RESUMEN

T cell antigen receptor (TCR) signaling triggers selective cytokine expression to drive T cell proliferation and differentiation required for immune defense and surveillance. The nuclear signaling events responsible for specificity in cytokine gene expression upon T cell activation are largely unknown. Here, we uncover formation of a dynamic actin filament network in the nucleus that regulates cytokine expression for effector functions of CD4+ T lymphocytes. TCR engagement triggers the rapid and transient formation of a nuclear actin filament network via nuclear Arp2/3 complex, induced by elevated nuclear Ca2+ levels and regulated via N-Wasp and NIK. Specific interference with TCR-induced formation of nuclear actin filaments impairs production of effector cytokines and prevents generation of antigen-specific antibodies but does not interfere with immune synapse formation and cell proliferation. Ca2+-regulated actin polymerization in the nucleus allows CD4+ T cells the rapid conversion of TCR signals into effector functions required for T cell help.


Asunto(s)
Actinas/inmunología , Antígenos/inmunología , Linfocitos T CD4-Positivos/inmunología , Receptores de Antígenos de Linfocitos T/inmunología , Complejo 2-3 Proteico Relacionado con la Actina/inmunología , Traslado Adoptivo , Animales , Línea Celular , Humanos , Ratones
2.
Rofo ; 185(7): 621-7, 2013 Jul.
Artículo en Alemán | MEDLINE | ID: mdl-23450372

RESUMEN

Iron as an essential nutrient is involved in multiple metabolic activities. The importance of a sufficient iron supply is stressed by the fact that, according to WHO data, about 30 % of the global population suffers from iron deficiency and resulting anemia. In contrast, hereditary hemochromatosis is the most common monogeneous inherited disease (prevalence of homozygous genotype 1:200 - 300 in Germany). While iron-induced anemia can be handled by relatively simple diagnostic and therapeutic management, the diagnosis and quantification of organ iron overload is far more challenging. This is of great clinical impact, as the overall body and organ iron concentration is the crucial prognostic parameter in iron overload disease. In 2001 the international workshop of NIDDK (National Institute of Diabetes and Digestive and Kidney Diseases) concluded that a quantitative, noninvasive, safe, and accurate approach for the assessment of body iron storage is needed to improve the diagnosis and management of patients with iron overload.


Asunto(s)
Algoritmos , Sobrecarga de Hierro/diagnóstico , Sobrecarga de Hierro/metabolismo , Hierro/análisis , Imagen por Resonancia Magnética/métodos , Espectroscopía de Resonancia Magnética/métodos , Humanos , Distribución Tisular
3.
Oncogene ; 32(44): 5241-52, 2013 Oct 31.
Artículo en Inglés | MEDLINE | ID: mdl-23208498

RESUMEN

Besides its established functions in intermediary metabolism and developmental processes, the nuclear receptor peroxisome proliferator-activated receptor ß/δ (PPARß/δ) has a less defined role in tumorigenesis. In the present study, we have identified a function for PPARß/δ in cancer cell invasion. We show that two structurally divergent inhibitory ligands for PPARß/δ, the inverse agonists ST247 and DG172, strongly inhibit the serum- and transforming growth factor ß (TGFß)-induced invasion of MDA-MB-231 human breast cancer cells into a three-dimensional matrigel matrix. To elucidate the molecular basis of this finding, we performed chromatin immunoprecipitation sequencing (ChIP-Seq) and microarray analyses, which identified the gene encoding angiopoietin-like 4 (ANGPTL4) as the major transcriptional PPARß/δ target in MDA-MB-231 cells, previously implicated in TGFß-mediated tumor progression and metastatic dissemination. We show that the induction of ANGPTL4 by TGFß and other oncogenic signals is strongly repressed by ST247 and DG172 in a PPARß/δ-dependent fashion, resulting in the inhibition of ANGPTL4 secretion. This effect is attributable to these ligands' ability to induce a dominant transcriptional repressor complex at the site of transcription initiation that blocks preinitiation complex formation through an histone deacetylase-independent, non-canonical mechanism. Repression of ANGPTL4 transcription by inverse PPARß/δ agonists is functionally linked to the inhibition of cancer cell invasion into a three-dimensional matrix, as (i) invasion of MDA-MB-231 cells is critically dependent on ANGPTL4 expression, (ii) recombinant ANGPTL4 stimulates invasion, and (iii) reverses the inhibitory effect of ST247 and DG172. These findings indicate that a PPARß/δ-ANGPTL4 pathway is involved in the regulation of tumor cell invasion and that its pharmacological manipulation by inverse PPARß/δ agonists is feasible.


Asunto(s)
Acrilonitrilo/análogos & derivados , Angiopoyetinas/genética , PPAR delta/fisiología , Piperazinas/farmacología , Transducción de Señal , Sulfonamidas/farmacología , Tiofenos/farmacología , Acrilonitrilo/farmacología , Proteína 4 Similar a la Angiopoyetina , Angiopoyetinas/metabolismo , Sitios de Unión , Línea Celular Tumoral , Movimiento Celular , Regulación Neoplásica de la Expresión Génica , Histona Desacetilasas/metabolismo , Humanos , Invasividad Neoplásica , PPAR delta/agonistas , Receptores X Retinoide/metabolismo , Iniciación de la Transcripción Genética/efectos de los fármacos , Factor de Crecimiento Transformador beta/fisiología
4.
Klin Padiatr ; 222(6): 399-406, 2010 Nov.
Artículo en Alemán | MEDLINE | ID: mdl-20862634

RESUMEN

In Germany and Central Europe, congenital disorders leading to secondary hemochromatosis are rare. The majority of these patients are treated in peripheral medical institutions. As a consequence, the experience of each institution in the treatment of secondary hemochromatosis in patients with congenital anemia is limited. Recent developments concerning new chelating agents, their combination for intensified chelation and new possibilities to diagnose and monitor iron overload have important consequences for the management of patients with secondary hemochromatosis and increase its complexity enormously. Therefore, the development of a guideline for rational and efficient diagnostics and treatment was necessary. The new guideline was developed within a formal consensus process and finally approved by a consensus conference with participants from both the pediatric and adult German hematology societies (GPOH and DGHO). Apart from general information and recommendations, the guideline contains 9 consensus statements on diagnostics (iron status, siderotic complications, chelator side-effects), the start of chelation, indications for intensified chelation, iron elimination in specific disorders, and iron elimination after stem cell transplantation. Here, these consensus statements are presented and discussed in detail. For the complete text of the guideline, please visit the AWMF homepage at http://www.leitlinien.net .


Asunto(s)
Anemia Aplásica/terapia , Anemia Diseritropoyética Congénita/terapia , Quelantes/uso terapéutico , Transfusión de Eritrocitos/efectos adversos , Hemocromatosis/tratamiento farmacológico , Hemosiderosis/tratamiento farmacológico , Anemia Aplásica/sangre , Anemia de Diamond-Blackfan/sangre , Anemia de Diamond-Blackfan/terapia , Anemia Diseritropoyética Congénita/sangre , Anemia de Células Falciformes/sangre , Anemia de Células Falciformes/terapia , Quelantes/efectos adversos , Niño , Deferoxamina/efectos adversos , Deferoxamina/uso terapéutico , Ferritinas/sangre , Alemania , Trasplante de Células Madre Hematopoyéticas/efectos adversos , Hemocromatosis/sangre , Hemocromatosis/diagnóstico , Hemosiderosis/sangre , Hemosiderosis/diagnóstico , Humanos , Talasemia beta/sangre , Talasemia beta/terapia
5.
Oncogene ; 29(16): 2441-8, 2010 Apr 22.
Artículo en Inglés | MEDLINE | ID: mdl-20101212

RESUMEN

Invasive cell migration is a key step for cancer metastasis and involves Rho GTPase-controlled reorganization of the actin cytoskeleton. Altered Rho GTPase expression is found in various malignancies. Particularly, the closely related GTPases RhoA and RhoC are upregulated in many aggressive tumours, but specific effectors that distinguish between these two GTPases to explain mechanistic differences have not been identified. The formins are by far the largest family of Rho GTPase effectors and are characterized by the actin-nucleating formin homology 2 domain. Using siRNA-based screening against all 15 human formins, we systematically analysed their functions in 3D cell motility using three different cancer cell lines. These results reveal distinct requirements for specific formins in amoeboid versus mesenchymal invasive cell migration. Importantly, by knocking down all Rho proteins, we identified formin-like 2 (FMNL2) as a specific RhoC effector, showing selective interaction of FMNL2 with active RhoC, but not RhoA or RhoB. Functional analysis shows that RhoC regulates autoinhibition of FMNL2, whereas suppression of FMNL2 inhibits RhoC-, but not RhoA-dependent, rounded invasive cell migration. Thus, our data uncover a novel regulatory and functional interaction between RhoC and FMNL2 for modulating cell shape and invasiveness and provide mechanistic insight into RhoC-specific signalling events.


Asunto(s)
Invasividad Neoplásica , Proteínas/fisiología , Proteínas de Unión al GTP rho/fisiología , Línea Celular Tumoral , Movimiento Celular , Forminas , Humanos , Proteínas/antagonistas & inhibidores , Proteínas/genética , Proteína rhoC de Unión a GTP
6.
Oncogene ; 28(7): 983-93, 2009 Feb 19.
Artículo en Inglés | MEDLINE | ID: mdl-19079344

RESUMEN

Transforming growth factor beta-1 (TGF-beta) acts as both a tumour suppressor and a tumour promoter in a context-dependent manner. The tumour-promoting activities of TGF-beta are likely to result from a combination of Smad and non-Smad signalling pathways but remain poorly understood. Here we show that TGF-beta-mediated activation of RhoA is dependent on the kinase activity of ALK5 and that continuous ALK5 activity maintains basal RhoA-ROCK signalling, cell morphology and actin dynamics in serum-starved rodent fibroblasts independently of Smad2, Smad3 and Smad4. In immortalized human diploid fibroblasts, we show that oncogenic rewiring by transduction of (V12)HaRas instigates regulation of RhoA-ROCK signalling through an autocrine TGF-beta1-ALK5 pathway. Furthermore, we show that ALK5-mediated activation of RhoA is required for efficient (V12)HaRas, V-Raf and (V600E)BRAF transformation and (V12)HaRas-mediated anchorage-independent growth. These findings identify a new pro-oncogenic activity of TGF-beta and indicate that tumours harbouring (V12)HaRas and (V600E)BRAF mutations may be susceptible to TGF-beta signalling inhibitors.


Asunto(s)
Transformación Celular Neoplásica/genética , Genes ras/fisiología , Proteínas Proto-Oncogénicas B-raf/genética , Transducción de Señal , Factor de Crecimiento Transformador beta/farmacología , Proteína de Unión al GTP rhoA/genética , Actinas/metabolismo , Animales , Benzamidas/farmacología , Western Blotting , Transformación Celular Neoplásica/metabolismo , Células Cultivadas , Citoesqueleto , Dioxoles/farmacología , Ensayo de Inmunoadsorción Enzimática , Fibroblastos/citología , Fibroblastos/metabolismo , Técnica del Anticuerpo Fluorescente , Guanosina Trifosfato/metabolismo , Humanos , Ratones , Células 3T3 NIH , Proteínas Serina-Treonina Quinasas/antagonistas & inhibidores , Proteínas Serina-Treonina Quinasas/metabolismo , Proteínas Proto-Oncogénicas B-raf/metabolismo , Ratas , Receptor Tipo I de Factor de Crecimiento Transformador beta , Receptores de Factores de Crecimiento Transformadores beta/antagonistas & inhibidores , Receptores de Factores de Crecimiento Transformadores beta/metabolismo , Proteína smad3/metabolismo , Proteína Smad4/metabolismo , Transfección , Quinasas Asociadas a rho/metabolismo , Proteína de Unión al GTP rhoA/metabolismo
7.
Am J Transplant ; 8(9): 1786-97, 2008 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-18786225

RESUMEN

Mycophenolic acid (MPA) is a potent inhibitor of the inosine monophosphate dehydrogenase and used as an immunosuppressive drug in transplantation. MPA inhibits proliferation of T- and B-lymphocytes by guanosine depletion. Since fibroblasts rely on the de novo synthesis of guanosine nucleotides, it is assumed that MPA interacts with fibroblasts causing an increased frequency of wound healing problems. We show a downregulation of the cytoskeletal proteins vinculin, actin and tubulin in fibroblasts exposed to pharmacological doses of MPA using microarray technology, real-time polymerase chain reaction (PCR) and Western blot. This reduction in RNA and protein content is accompanied by a substantial rearrangement of the cytoskeleton in MPA-treated fibroblasts as documented by immunofluorescence. The dysfunctional fibroblast growth was validated by scratch test documenting impaired migrational capacity. In contrast, cell adhesion was increased in MPA-treated fibroblasts. The results of the cultured human fibroblasts were applied to skin biopsies of renal transplant recipients. Skin biopsies of patients treated with MPA expressed less vinculin, actin and tubulin as compared to control biopsies that could explain potential wound healing problems posttransplantation. The perspective of MPA-induced cytoskeletal dysfunction may go beyond wound healing disturbances and may have beneficial effects on (renal) allografts with respect to scarring.


Asunto(s)
Movimiento Celular/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Fibroblastos/efectos de los fármacos , Inmunosupresores/farmacología , Ácido Micofenólico/farmacología , Biopsia , Carbocianinas/metabolismo , Adhesión Celular/efectos de los fármacos , Células Cultivadas , Procedimientos Quirúrgicos Dermatologicos , Relación Dosis-Respuesta a Droga , Fluoresceína-5-Isotiocianato/metabolismo , Colorantes Fluorescentes/metabolismo , Humanos , Inmunohistoquímica , Indoles/metabolismo , Faloidina/metabolismo , Rodaminas/metabolismo , Piel/citología
9.
Oncogene ; 27(12): 1737-48, 2008 Mar 13.
Artículo en Inglés | MEDLINE | ID: mdl-17906699

RESUMEN

Neuropeptide hormones like bombesin/gastrin-releasing peptide, galanin or bradykinin, acting via auto and paracrine growth loops, represent the principal mitogens of small cell lung cancer (SCLC). These mitogenic neuropeptides activate G(q/11)-coupled receptors which stimulate phospholipase Cbeta activity, followed by rises of the intracellular calcium concentration ([Ca2+](i)) and activation of protein kinase C (PKC). We report here that proline-rich tyrosine kinase Pyk2 is highly expressed in SCLC cells and provides a functional link between neuropeptide-induced increases in [Ca2+](i) and tumor cell proliferation. Activation of Pyk2 and its association with Src kinases critically depends on the elevation of [Ca2+](i), but is independent of PKC. Src kinase activities are crucial for neuropeptide-mediated GTP-loading of Ras and activation of extracellular signal-regulated kinases in SCLC cells. Pyk2 and Src kinases essentially contribute to anchorage-independent proliferation of SCLC cells. Inhibition of either Pyk2 or Src kinases by lentiviral RNAi or pharmacological inhibition with PP2, respectively, attenuated basal and neuropeptide-elicited survival and proliferation of SCLC cells in liquid culture and in soft agar. Thus, neuropeptides stimulate anchorage-independent survival and proliferation of SCLC cells via pathways involving Pyk2 and Src kinases. Therefore, Ca2+-induced Pyk2/Src complex formation may be a rewarding molecular target for novel therapeutic strategies in SCLC cells.


Asunto(s)
Carcinoma de Células Pequeñas/enzimología , Proliferación Celular , Quinasa 2 de Adhesión Focal/fisiología , Galanina/fisiología , Neoplasias Pulmonares/enzimología , Familia-src Quinasas/fisiología , Calcio/fisiología , Carcinoma de Células Pequeñas/patología , Línea Celular , Línea Celular Tumoral , Activación Enzimática/genética , Quinasa 2 de Adhesión Focal/biosíntesis , Quinasa 2 de Adhesión Focal/genética , Quinasa 2 de Adhesión Focal/metabolismo , Humanos , Neoplasias Pulmonares/patología , Prolina/metabolismo , Familia-src Quinasas/metabolismo
10.
Clin Exp Allergy ; 37(11): 1701-8, 2007 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-17892512

RESUMEN

BACKGROUND: Neurotrophins are involved in inflammatory reactions influencing several cells in health and disease including allergy and asthma. Dendritic cells (DCs) play a major role in the induction of inflammatory processes with an increasing role in allergic diseases as well. OBJECTIVE: The aim of this study was to investigate the influence of neurotrophins on DC function. METHODS: Monocyte-derived dendritic cells were generated from allergic and non-allergic donors. Neurotrophin receptors were demonstrated by western blotting, flow cytometry and fluorescence microscopy. Activation of small GTPases was evaluated by pull-down assays. DCs were incubated with nerve growth factor (NGF) and brain-derived neurotrophic factor (BDNF) and supernatants were collected for measurement of IL-4, IL-6, IL-10, IL-12p70, TNF-alpha and TGF-beta. RESULTS: Receptor proteins were detectable by western blot, fluorescence activated cell sorting analysis and fluorescence microscopy. Signalling after neurotrophin stimulation occurred in a ligand-specific pattern. NGF led to decreased RhoA and increased Rac activation, while BDNF affected RhoA and Rac activity in a reciprocal fashion. Cells of allergics released a significantly increased amount of IL-6, while for healthy subjects a significantly higher amount of IL-10 was found. CONCLUSION: These data indicate that DCs are activated by the neurotrophins NGF and BDNF by different pathways in a receptor-dependant manner. These cells then may initiate inflammatory responses based on allergic sensitization releasing preferred cytokines inducing tolerance or a T-helper type 2 response.


Asunto(s)
Factor Neurotrófico Derivado del Encéfalo/farmacología , Células Dendríticas/efectos de los fármacos , Factor de Crecimiento Nervioso/farmacología , Antígenos CD/metabolismo , Western Blotting , Ligando de CD40/farmacología , Diferenciación Celular/efectos de los fármacos , Células Cultivadas , Citocinas/farmacología , Células Dendríticas/citología , Células Dendríticas/metabolismo , Relación Dosis-Respuesta a Droga , Citometría de Flujo , Expresión Génica/efectos de los fármacos , Humanos , Hipersensibilidad/sangre , Interleucina-10/metabolismo , Interleucina-6/metabolismo , Lipopolisacáridos/farmacología , Monocitos/citología , Monocitos/efectos de los fármacos , Monocitos/metabolismo , Poli I-C/farmacología , Receptor trkA/genética , Receptor trkA/metabolismo , Receptor trkB/genética , Receptor trkB/metabolismo , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Linfocitos T/citología , Linfocitos T/efectos de los fármacos , Linfocitos T/metabolismo , Proteínas de Unión al GTP rac/metabolismo , Proteínas de Unión al GTP rho/metabolismo
11.
Ann Oncol ; 17(8): 1205-12, 2006 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-16766587

RESUMEN

BACKGROUND: The aim of the study was to analyse the toxicity and health related quality of life (HRQoL) of breast cancer patients treated with FAC (5-fluorouracil, doxorubicin, cyclophosphamide) and TAC (docetaxel, doxorubicin, cyclophosphamide) with and without primary prophylactic G-CSF (PPG). PATIENTS AND METHODS: This was a phase III study to compare FAC and TAC as adjuvant treatment of high-risk node-negative breast cancer patients. After the entry of the first 237 patients, the protocol was amended to include PPG in the TAC arm due to the high incidence of febrile neutropenia. A total of 1047 evaluable patients from 49 centres in Spain, two in Poland and four in Germany were included in the trial. Side-effects and the scores of the EORTC QLQ-C30 and QLQ BR-23 questionnaires were compared in the three groups (FAC, TAC pre-amendment and TAC post-amendment). RESULTS: The addition of PPG to TAC significantly reduced the incidence of neutropenic fever, grade 2-4 anaemia, asthenia, anorexia, nail disorders, stomatitis, myalgia and dysgeusia. Patient QoL decreased during chemotherapy, more with TAC than FAC, but returned to baseline values afterwards. The addition of PPG to TAC significantly reduced the percentage of patients with clinically relevant Global Health Status deterioration (10 or more points over baseline value) at the end of chemotherapy (64% versus 46%, P<0.03). CONCLUSIONS: The addition of PPG significantly reduces the incidence of neutropenic fever associated with TAC chemotherapy as well as that of some TAC-induced haematological and extrahaematological side-effects. The HRQoL of patients treated with TAC is worse than that of those treated with FAC but improves with the addition of PPG, particularly in the final part of chemotherapy treatment.


Asunto(s)
Protocolos de Quimioterapia Combinada Antineoplásica/efectos adversos , Neoplasias de la Mama/tratamiento farmacológico , Factor Estimulante de Colonias de Granulocitos/uso terapéutico , Neutropenia/prevención & control , Adolescente , Adulto , Anciano , Protocolos de Quimioterapia Combinada Antineoplásica/uso terapéutico , Quimioterapia Adyuvante , Ciclofosfamida/administración & dosificación , Ciclofosfamida/efectos adversos , Ciclofosfamida/uso terapéutico , Docetaxel , Doxorrubicina/administración & dosificación , Doxorrubicina/efectos adversos , Doxorrubicina/uso terapéutico , Femenino , Fluorouracilo/administración & dosificación , Fluorouracilo/efectos adversos , Fluorouracilo/uso terapéutico , Enfermedades Hematológicas/inducido químicamente , Enfermedades Hematológicas/prevención & control , Humanos , Persona de Mediana Edad , Neutropenia/inducido químicamente , Calidad de Vida , Taxoides/administración & dosificación , Taxoides/efectos adversos , Taxoides/uso terapéutico
12.
Eur Respir J ; 19(2): 374-6, 2002 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-11866018

RESUMEN

By using a combination of multiplex polymerase chain reaction and allele-specific labelled probes, the oligo-ligation assay is designed to detect known cystic fibrosis transmembrane regulator mutations. This study shows that this assay may also be useful to detect new mutations. The second child of a family of Bosnic origin showed all the symptoms of intestinal and pulmonary manifestations of cystic fibrosis. No signal could be obtained for the allele-specific probe 1898+1G>A. This could be explained by a nearby localized sequence change that prevented polymerase chain reaction primers or oligonucleotide probes from binding to the target sequence. Indeed, sequence analysis revealed a new 1894G>T exchange (Glu587Stop). Both parents and the healthy brother carried this mutation. Thus, the index patient was homozygous for 1894G>T, which was inherited from both parents.


Asunto(s)
Codón sin Sentido , Regulador de Conductancia de Transmembrana de Fibrosis Quística/genética , Fibrosis Quística/genética , Bosnia y Herzegovina , Genética de Población , Homocigoto , Humanos , Lactante , Reacción en Cadena de la Polimerasa
13.
Oncogene ; 19(37): 4199-209, 2000 Aug 31.
Artículo en Inglés | MEDLINE | ID: mdl-10980593

RESUMEN

Neuropeptides like galanin produced and released by small cell lung cancer (SCLC) cells are considered principal mitogens in these tumors. We identified the galanin receptor type 2 (GALR2) as the only galanin receptor expressed in H69 and H510 cells. Photoaffinity labeling of G proteins in H69 cell membranes revealed that GALR2 activates G proteins of three subfamilies: G(q), G(i), and G(12). In H69 cells, galanin-induced Ca2+ mobilization was pertussis toxin-insensitive. While phorbol ester-induced extracellular signal-regulated kinase (ERK) activation required protein kinase C (PKC) activity, preincubation of H69 cells with the PKC-inhibitor GF109203X had no effect on galanin-dependent ERK activity. A rise of the intracellular calcium concentration was necessary and sufficient to mediate galanin-induced ERK activation. In support of G(i) coupling, stimulation of GALR2 expressed in HEK293 cells inhibited isoproterenol-induced cAMP accumulation and raised cAMP levels in COS-7 cells when coexpressed with a chimeric G alpha(S)-G alpha(i) protein In H69 cells, galanin activated the monomeric GTPase RhoA and induced stress fiber formation in Swiss 3T3 cells expressing GALR2. Thus, we provide the first direct evidence that in SCLC the mitogenic neuropeptide galanin, interacting with GALR2, simultaneously activates multiple classes of G proteins and signals through the G(q) phospholipase C/calcium sequence and a G(12)/Rho pathway. Oncogene (2000) 19, 4199 - 4209


Asunto(s)
Señalización del Calcio/fisiología , Carcinoma de Células Pequeñas/patología , Subunidades alfa de la Proteína de Unión al GTP Gi-Go/fisiología , Proteínas de Unión al GTP/fisiología , Proteínas de Unión al GTP Heterotriméricas/fisiología , Neoplasias Pulmonares/patología , Proteínas de Neoplasias/fisiología , Receptores de Neuropéptido/fisiología , 1-Metil-3-Isobutilxantina/farmacología , Células 3T3/efectos de los fármacos , Citoesqueleto de Actina/metabolismo , Actinas/metabolismo , Animales , Células COS/efectos de los fármacos , Señalización del Calcio/efectos de los fármacos , Carcinoma de Células Pequeñas/metabolismo , Adhesión Celular , AMP Cíclico/fisiología , Inhibidores Enzimáticos/farmacología , Subunidades alfa de la Proteína de Unión al GTP G12-G13 , Subunidades alfa de la Proteína de Unión al GTP Gq-G11 , Galanina/metabolismo , Galanina/farmacología , Humanos , Indoles/farmacología , Fosfatos de Inositol/metabolismo , Péptidos y Proteínas de Señalización Intracelular , Isoproterenol/farmacología , Neoplasias Pulmonares/metabolismo , Maleimidas/farmacología , Ratones , Proteína Quinasa 1 Activada por Mitógenos/metabolismo , Proteína Quinasa 3 Activada por Mitógenos , Proteínas Quinasas Activadas por Mitógenos/metabolismo , Proteínas de Neoplasias/antagonistas & inhibidores , Toxina del Pertussis , Fosfatidilinositol Diacilglicerol-Liasa , Proteína Quinasa C/antagonistas & inhibidores , Proteína Quinasa C/fisiología , Proteínas Serina-Treonina Quinasas/fisiología , Receptor de Galanina Tipo 2 , Receptores de Galanina , Receptores de Neuropéptido/efectos de los fármacos , Proteínas Recombinantes de Fusión/genética , Proteínas Recombinantes de Fusión/fisiología , Transfección , Células Tumorales Cultivadas/metabolismo , Fosfolipasas de Tipo C/metabolismo , Factores de Virulencia de Bordetella/farmacología , Quinasas Asociadas a rho , Proteína de Unión al GTP rhoA/fisiología
14.
J Biol Chem ; 275(16): 12251-60, 2000 Apr 21.
Artículo en Inglés | MEDLINE | ID: mdl-10766863

RESUMEN

Gonadotropin releasing hormone (GnRH) contributes to the maintenance of gonadotrope function by increasing extracellular signal-regulated kinase (ERK) activity subsequent to binding to its cognate G-protein-coupled receptor. As the GnRH receptor exclusively interacts with G(q/11) proteins and as receptor expression is regulated in a beta-arrestin-independent fashion, it represents a good model to systematically dissect underlying signaling pathways. In alphaT3-1 gonadotropes endogenously expressing the GnRH receptor, GnRH challenge resulted in a rapid increase in ERK activity which was attenuated by the epidermal growth factor receptor (EGFR)-specific tyrosine kinase inhibitor AG1478. In COS-7 cells transiently expressing the human GnRH receptor, agonist-induced ERK activation was independent of free Gbetagamma subunits but could be mimicked by short-term phorbol ester treatment. Most notably, G(q/11)-induced ERK activation was sensitive to N17-Ras and to expression of the C-terminal Src kinase but also to other dominant negative mutants of signaling components localized upstream of Ras, like Shc and the EGFR. GnRH as well as phorbol esters led to Ras activation in COS-7 and alphaT3-1 cells, which was dependent on Src and EGFR tyrosine kinases, indicating that both tyrosine kinases act downstream of protein kinase C (PKC) and upstream of Ras. However, Src did not contribute to Shc tyrosine phosphorylation. GnRH or phorbol ester challenge resulted in PKC-dependent EGFR autophosphorylation. Furthermore, a 5-min phorbol ester treatment was sufficient to trigger tyrosine phosphorylation of the platelet-derived growth factor-beta receptor in L cells. Thus, in several cell systems PKC is able to stimulate Ras via activation of receptor tyrosine kinases.


Asunto(s)
Receptores ErbB/fisiología , Activadores de GTP Fosfohidrolasa/metabolismo , Hormona Liberadora de Gonadotropina/fisiología , Proteínas Quinasas Activadas por Mitógenos/metabolismo , Proteínas Activadoras de ras GTPasa/fisiología , Animales , Células COS , Calcio/metabolismo , Electroforesis en Gel de Poliacrilamida , Activación Enzimática , Humanos , Células L , Ratones , Proteínas Quinasas Activadas por Mitógenos/genética , Fosforilación , Adenohipófisis/metabolismo , Proteína Quinasa C/metabolismo , Proteínas Tirosina Quinasas Receptoras/genética , Proteínas Tirosina Quinasas Receptoras/metabolismo , Transfección
15.
Naunyn Schmiedebergs Arch Pharmacol ; 361(4): 345-62, 2000 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-10763849

RESUMEN

While the effects of receptor/G protein systems on intermediary metabolism have been intensively studied, it has only recently been appreciated that G protein-coupled receptors and G proteins (heterotrimeric GTP-binding proteins) play important roles in the regulation of cell growth, differentiation and even transformation. Naturally occurring mutations both in G protein-coupled receptors and in G protein alpha-subunits lead to autonomous cell growth resulting in human disease. One mechanism to transduce mitogenic signals from the cell membrane to the nucleus is the engagement of the extracellular signal-regulated kinase (ERK)mitogen-activated protein kinase (MAPK) cascade. Multiple distinct signal transduction pathways have been characterized which link G proteins with the ERK cascade. Receptor and non-receptor tyrosine kinases play central roles in these pathways. Mitogenic signaling by receptor/G protein systems is realized as a complex interplay between signals emanating from different classes of cell surface receptors. The characterization of receptor-, G protein- and tyrosine kinase-specific contributions to mitogenic signaling in a particular cell may ultimately allow for the rational design and application of pharmaceuticals to treat diseases involving uncontrolled cell proliferation.


Asunto(s)
Proteínas de Unión al GTP/fisiología , Receptores de Superficie Celular/fisiología , Transducción de Señal/fisiología , Animales , División Celular/fisiología , Transformación Celular Neoplásica , Proteínas de Unión al GTP/genética , Proteínas de Unión al GTP Heterotriméricas/genética , Proteínas de Unión al GTP Heterotriméricas/fisiología , Humanos , Proteínas Quinasas Activadas por Mitógenos/metabolismo , Proteínas Quinasas Activadas por Mitógenos/fisiología , Mutación
16.
J Biol Chem ; 275(13): 9193-200, 2000 Mar 31.
Artículo en Inglés | MEDLINE | ID: mdl-10734055

RESUMEN

The agonist-bound gonadotropin-releasing hormone (GnRH) receptor engages several distinct signaling cascades, and it has recently been proposed that coupling of a single type of receptor to multiple G proteins (G(q), G(s), and G(i)) is responsible for this behavior. GnRH-dependent signaling was studied in gonadotropic alphaT3-1 cells endogenously expressing the murine receptor and in CHO-K1 (CHO#3) and COS-7 cells transfected with the human GnRH receptor cDNA. In all cell systems studied, GnRH-induced phospholipase C activation and Ca(2+) mobilization was pertussis toxin-insensitive, as was GnRH-mediated extracellular signal-regulated kinase activation. Whereas the G(i)-coupled m2 muscarinic receptor interacted with a chimeric G(s) protein (G(s)i5) containing the C-terminal five amino acids of Galpha(i2), the human GnRH receptor was unable to activate the G protein chimera. GnRH challenge of alphaT3-1, CHO#3 and of GnRH receptor-expressing COS-7 cells did not result in agonist-dependent cAMP formation. GnRH challenge of CHO#3 cells expressing a cAMP-responsive element-driven firefly luciferase did not result in increased reporter gene expression. However, coexpression of the human GnRH receptor and adenylyl cyclase I in COS-7 cells led to clearly discernible GnRH-dependent cAMP formation subsequent to GnRH-elicited rises in [Ca(2+)](i). In alphaT3-1 and CHO#3 cell membranes, addition of [alpha-(32)P]GTP azidoanilide resulted in GnRH receptor-dependent labeling of Galpha(q/11) but not of Galpha(i), Galpha(s) or Galpha(12/13) proteins. Thus, the murine and human GnRH receptors exclusively couple to G proteins of the G(q/11) family. Multiple GnRH-dependent signaling pathways are therefore initiated downstream of the receptor/G protein interface and are not indicative of a multiple G protein coupling potential of the GnRH receptor.


Asunto(s)
Proteínas de Unión al GTP/metabolismo , Receptores LHRH/metabolismo , Transducción de Señal , Adenilil Ciclasas/metabolismo , Animales , Células CHO , Células COS , Calmodulina/metabolismo , Cricetinae , AMP Cíclico/biosíntesis , Humanos , Mutagénesis , Unión Proteica , Receptores LHRH/genética , Proteínas Recombinantes de Fusión/genética , Proteínas Recombinantes de Fusión/metabolismo
17.
Int J Cancer ; 85(4): 474-81, 2000 Feb 15.
Artículo en Inglés | MEDLINE | ID: mdl-10699917

RESUMEN

We have developed a 2-stage protocol for BRCA1 and BRCA2 mutation screening from blood spot paper. Stage 1 screening was aimed to analyze patients at highest risk for the most common disease-associated sequence variants listed in the BIC database. Accordingly, stage1 testing implied detection of 18 disease- associated BRCA1 and 9 BRCA2 mutations by adapting the 5' nuclease assay to heterozygote screening. For stage 2 screening, we applied the conformation sensitive gel electrophoresis (CSGE) method by adapting this technique to automated heteroduplex analysis of BRCA1 and BRCA2 using fragment scanning on an ABI 377 sequencing device. Of the 120 patients with a family history of breast and ovarian cancer who took part in this study so far, 45 entered stage 1 testing. Disease-associated mutations were detected in 6 patients by stage 1 testing (13%). For these patients, the final result was available within 10 days. Mutation 300T-->G was found in 2 patients. One patient with mutation 3036delACAA in BRCA2 reported only 1 sister with a multifocal bilateral breast cancer. New disease-associated mutations were detected in 2 of the 114 patients who entered the stage 2 test (1.7%). Of particular interest was 1 patient who was diagnosed with a medullary breast carcinoma at age 39 and who had no family history of breast cancer. We conclude that pre-screening by 5' nuclease assay for the mutations most frequently seen in a given population represents a relatively effective first line of analysis. Subsequent detailed analysis by fluorescence conformation sensitive gel electrophoresis (F-CSGE) and fragment sequencing is a sensitive alternative to full nucleotide sequencing.


Asunto(s)
Neoplasias de la Mama/genética , Genes BRCA1 , Genes Supresores de Tumor , Mutación , Proteínas de Neoplasias/genética , Neoplasias Ováricas/genética , Factores de Transcripción/genética , Adulto , Edad de Inicio , Anciano , Proteína BRCA2 , Exones , Familia , Femenino , Pruebas Genéticas , Humanos , Intrones , Pérdida de Heterocigocidad , Persona de Mediana Edad , Mutación Puntual , Eliminación de Secuencia
18.
J Biol Chem ; 275(4): 2381-9, 2000 Jan 28.
Artículo en Inglés | MEDLINE | ID: mdl-10644689

RESUMEN

Previous studies have established that G-protein-coupled receptors (GPCRs) are composed of independent folding domains. Based on this findings we attempted to rescue the function of clinically relevant missense mutations (R137H, S167L, and R181C) within the N-terminal domain of the V2 vasopressin receptor (V2-R), by coexpressing mutated full-length (Y280C) and C-terminally truncated (E242X) receptor constructs in COS-7 cells. Coimmunoprecipitation and enzyme-linked immunosorbent assay studies demonstrated a specific association of E242X with full-length V2-Rs even in the presence of missense mutations. Systematic analysis of the structural requirements for the observed receptor/fragment association showed that N-terminal fragments containing at least transmembrane regions 1-3 interact with the full-length V2-R. Despite this specific interaction, no functional reconstitution was achieved for mutant V2-Rs following coexpression with E242X and Y280C. However, functional activity of R137H and R181C upon coexpression with E242X was regained by mutational disruption of the extracellular disulfide bond, which is highly conserved among GPCRs. Our data with the V2-R are consistent with a structural model in which class I GPCRs form contact oligomers by lateral interaction rather than by a domain-swapping mechanism.


Asunto(s)
Receptores de Vasopresinas/química , Secuencia de Aminoácidos , Animales , Biopolímeros , Células COS , Disulfuros/química , Datos de Secuencia Molecular , Mutagénesis , Receptores de Vasopresinas/genética
19.
Biochim Biophys Acta ; 1446(1-2): 57-70, 1999 Jul 07.
Artículo en Inglés | MEDLINE | ID: mdl-10395919

RESUMEN

Based on structural similarities of an expressed sequence tag with the platelet-activating factor (PAF) receptor a cDNA clone encoding a novel G-protein-coupled receptor (GPCR), named GPR34, was isolated from a human fetal brain cDNA library. Genomic DNA analyses revealed the receptor to be encoded by an intronless single-copy gene at Xp11. 3-11.4. The predicted 381-amino-acid protein disclosed all structural features characteristic of a member of the class I GPCR family. Except an obvious sequence homology in transmembrane domain 6, no further similarities to the PAF receptor or any other known GPCR were found. The corresponding mouse receptor DNA was isolated from a genomic P1 library displaying a 90% amino acid identity compared to the human receptor. Phylogenetic studies showed that GPR34 is preserved among vertebrates, and the existence of GPR34 subtypes was demonstrated. The receptor mRNA is abundantly expressed in human and mouse tissues. In addition to the major 2-kb transcript, a 4-kb transcript was found only in mouse liver and testis. Expression of the human GPR34 in COS-7 cells followed by Western blot studies revealed specific bands of a highly glycosylated protein between 75 and 90 kDa. A number of potential ligands including phospholipids, leukotrienes, hydroxy-eicosatetraenoic acids, nucleotides and peptides were tested in functional assays. Since none of the applied substances led to significant changes in second messenger levels (cAMP and inositol phosphates), the natural ligand and coupling profile of this novel GPCR subgroup remains unknown.


Asunto(s)
Proteínas de Unión al GTP/metabolismo , Receptores de Superficie Celular/clasificación , Receptores Acoplados a Proteínas G , Secuencia de Aminoácidos , Animales , Secuencia de Bases , Encéfalo/embriología , Encéfalo/metabolismo , Células COS , Clonación Molecular , Regulación de la Expresión Génica , Biblioteca de Genes , Humanos , Ratones , Datos de Secuencia Molecular , Filogenia , Glicoproteínas de Membrana Plaquetaria/química , Glicoproteínas de Membrana Plaquetaria/genética , ARN Mensajero/análisis , Receptores de Superficie Celular/genética , Receptores de Superficie Celular/metabolismo , Receptores Lisofosfolípidos , Receptores Purinérgicos P2/química , Receptores Purinérgicos P2/genética , Alineación de Secuencia
20.
Biochem Soc Symp ; 63: 51-69, 1998.
Artículo en Inglés | MEDLINE | ID: mdl-9513711

RESUMEN

Mammary gland development is controlled by systemic hormones and by growth factors that might complement or mediate hormonal action and provide the signalling basis for mesenchyme-epithelial cross-talk. Two locally expressed factors, pleiotrophin and mammary-derived growth inhibitor (MDGI), their hormonal regulation and proposed functions will be discussed. Pleiotrophin expression in non-tumorigenic, attachment-dependent epithelial cells leads to an attachment-independent, highly tumorigenic phenotype. The fatty acid binding protein MDGI specifically inhibits growth of normal mouse mammary epithelial cells, whereas growth of stromal cells is not suppressed. In mammary gland organ culture, inhibition of ductal growth by MDGI is associated with the appearance of bulbous alveolar end buds and formation of fully developed lobulo-alveolar structures. In parallel, MDGI stimulates its own epithelial-restricted expression and promotes milk protein synthesis. Selective inhibition of endogenous MDGI expression suppresses the appearance of alveolar end buds and lowers the beta-casein level in organ cultures. MDGI activity can be antagonized by epidermal growth factor (EGF); reciprocally, MDGI can suppress the mitogenic effects of EGF. An MDGI-derived C-terminal 11-amino-acid peptide is able to mimic MDGI activity in vitro. In conclusion, members of the family of fatty acid binding proteins are able to regulate mammary gland differentiation locally, and fatty acid binding is not required for this activity.


Asunto(s)
Proteínas Portadoras/fisiología , Diferenciación Celular , Citocinas/fisiología , Inhibidores de Crecimiento , Glándulas Mamarias Animales/citología , Mitógenos , Secuencia de Aminoácidos , Animales , Proteínas Portadoras/química , Proteínas Portadoras/farmacología , Diferenciación Celular/efectos de los fármacos , División Celular/efectos de los fármacos , Citocinas/química , Citocinas/farmacología , Proteína 3 de Unión a Ácidos Grasos , Proteínas de Unión a Ácidos Grasos , Femenino , Datos de Secuencia Molecular
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...